WO2005013889A9 - Fragments tronques d'alpha-synucleine dans les affections des corps de lewy - Google Patents

Fragments tronques d'alpha-synucleine dans les affections des corps de lewy

Info

Publication number
WO2005013889A9
WO2005013889A9 PCT/US2004/015836 US2004015836W WO2005013889A9 WO 2005013889 A9 WO2005013889 A9 WO 2005013889A9 US 2004015836 W US2004015836 W US 2004015836W WO 2005013889 A9 WO2005013889 A9 WO 2005013889A9
Authority
WO
WIPO (PCT)
Prior art keywords
synuclein
alpha
fragment
lbd
amino acids
Prior art date
Application number
PCT/US2004/015836
Other languages
English (en)
Other versions
WO2005013889A2 (fr
WO2005013889A3 (fr
Inventor
Tamie J Chilcote
Jason Goldstein
John P Anderson
Original Assignee
Elan Pharm Inc
Tamie J Chilcote
Jason Goldstein
John P Anderson
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Elan Pharm Inc, Tamie J Chilcote, Jason Goldstein, John P Anderson filed Critical Elan Pharm Inc
Priority to EP04776059.0A priority Critical patent/EP1633189B1/fr
Priority to DK04776059.0T priority patent/DK1633189T3/en
Priority to SI200432403T priority patent/SI1633189T1/sl
Priority to ES04776059.0T priority patent/ES2640669T3/es
Priority to CA2526900A priority patent/CA2526900C/fr
Priority to JP2006514909A priority patent/JP4820291B2/ja
Priority to PL04776059T priority patent/PL1633189T3/pl
Priority to US10/969,335 priority patent/US7306945B2/en
Publication of WO2005013889A2 publication Critical patent/WO2005013889A2/fr
Publication of WO2005013889A3 publication Critical patent/WO2005013889A3/fr
Publication of WO2005013889A9 publication Critical patent/WO2005013889A9/fr
Priority to US11/194,115 priority patent/US7358331B2/en
Priority to US12/040,739 priority patent/US20090035217A1/en
Priority to US13/604,441 priority patent/US9879071B2/en
Priority to US15/466,543 priority patent/US20170204173A1/en
Priority to CY20171100989T priority patent/CY1119556T1/el
Priority to US15/981,008 priority patent/US20180355028A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1096Processes for the isolation, preparation or purification of DNA or RNA cDNA Synthesis; Subtracted cDNA library construction, e.g. RT, RT-PCR
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids

Definitions

  • LBDs Lewy body diseases
  • DLB Lewy body diseases
  • AD Alzheimer's disease
  • AD, PD, and DLBD are the most commonly found neurodegenerative disorders in the elderly. Recent epidemiological studies have demonstrated a close clinical relationship between AD and PD, as about 30% of Alzheimer's patients also have PD. Compared to the rest of the aging population, patients with AD are thus more likely to develop concomitant PD. Furthermore, PD patients that become demented usually have developed classical AD. Although each neurodegenerative disease appears to have a predilection for specific brain regions and cell populations, resulting in distinct pathological features, PD, AD, and DLBD also share common pathological hallmarks. Patients with familial AD, Down syndrome, or sporadic AD develop LBs on the amygdala, which are the classical neuropathological hallmarks of PD.
  • each disease is associated with the degeneration of neurons, interneuronal synaptic connections and eventually cell death, the depletion of neurotransmitters, and abnormal accumulation of misfolded proteins, the precursors of which participate in normal central nervous system function.
  • Biochemical studies have confirmed a link between AD, PD and DLB.
  • the neuritic plaques that are the classic pathological hallmark of AD consist essentially of amyloid beta (A ⁇ ) pep tide, an amino acid proteolytic product of the amyloid precursor protein (APP), and NAC, a 35 amino acid proteolytic fragment of alpha-synuclein.
  • a ⁇ and NAC were first identified in amyloid plaques as proteolytic fragments of their respective full-length proteins, for which the full-length cDNAs were identified and cloned. (Iwai A., Biochim. Biophys. Ada (2000) 1502:95-109); Masliah et al, AM. J Pathol (1996) 148:201-10; Ueda et al, Proc. Natl Acad. Sci. USA (1993) 90:11282-6).
  • Alpha-synuclein is part of a large family of proteins including beta- and gamma- synuclein and synoretin. Alpha-synuclein is expressed in the normal state associated with synapses and is believed to play a role in neural plasticity, learning and memory. Mutations in human (h) alpha-synuclein that enhance the aggregation of alpha-synuclein have been identified (Ala30Pro and Ala53Thr) and are associated with rare forms of autosomal dominant forms of PD. The mechanism by which these mutations increase the propensity of alpha-synuclein to aggregate are unknown. SUMMARY OF THE CLAIMED INVENTION
  • the invention provides methods of screening for an agent having a pharmacological activity useful for treating a Lewy Body Disease (LBD).
  • LBD Lewy Body Disease
  • the method involves contacting the agent with a fragment of alpha-synuclein, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; and determining the rate or extent of aggregation of the fragment of alpha-synuclein, wherein a reduction in the rate or extent of aggregation relative to a control lacking the agent indicates the agent has the pharmacological activity.
  • the fragment has a C-terminus at a residue between 118 and 125 of intact alpha-synuclein.
  • Preferred fragments include alpha-synuclein 1-119, 1-120, 1-121, 1- 122, 1-123, 1-124, and 1-125 of alpha synuclein.
  • the fragment of alpha-synuclein is 1-X, wherein X is 130-139.
  • the fragment of alpha-synuclein bears a mutation associated with a hereditary LBD, such as an A53T mutations.
  • the method involves an additional step of conducting a trial in a human having a LBD or an animal model of LBD to determine whether the agent treats or inhibits a symptom of the LBD.
  • the invention further provides methods of screening an agent for a pharmacological activity useful in treating a LBD (e.g., Parkinson's disease or DLBD). These methods comprise contacting a cell expressing alpha-synuclein and processing the alpha-synuclein into a fragment with an agent.
  • the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein.
  • the fragment of alpha- synuclein has a C-terminus at a residue between 118 and 125 of intact alpha-synuclein.
  • Preferred fragments are 1-119, 1-120, 1-121, 1-122, 1-123, 1-124, and 1-125 of alpha synuclein.
  • the fragment of alpha-synuclein is 1-X, wherein X is 130-139.
  • the fragment of alpha-synuclein bears a mutation associated with a hereditary LBD, such as an A53T mutations.
  • the cell can be a human cell, a neuronal cell or a dopaminergic cell.
  • the cell is a PC 12 or Sy5Y cell.
  • the method involves a step of conducting a trial in a human having a LBD or an animal model of LBD to determine whether the agent treats or inhibits a symptom of the LBD.
  • the invention further provides methods of screening for an agent having a pharmacological activity useful for treating a LBD (e.g., Parkinson's disease or DLBD).
  • the methods involve contacting a transgenic animal expressing a fragment of alpha-synuclein, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; and determining a level of aggregated forms of the fragment in the brain of the transgenic animal relative to a baseline level of aggregated forms of the fragment in a comparable transgenic animal in the absence of the agent, a reduction in the level of the aggregated forms fragment relative to the baseline indicating the agent has a pharmacological activity useful in treating a LBD.
  • a LBD e.g., Parkinson's disease or DLBD
  • the fragment of alpha-synuclein has a C- terminus at a residue between 118 and 125 of intact alpha-synuclein.
  • Preferred fragments include 1-119, 1-120, 1-121, 1-122, 1-123, 1-124, and 1-125 of alpha synuclein.
  • the fragment of alpha-synuclein is 1-X, wherein X is 119-139.
  • the fragment of alpha-synuclein bears a mutation associated with a hereditary LBD, such as an A53T mutation.
  • the transgenic animal is a rodent.
  • the transgenic animal can also be a Drosophila.
  • the method involves conducting a trial in a human having a LBD or an animal model of LBD to determine whether the agent treats or inhibits a symptom of the LBD.
  • the invention further provides methods of screening an agent for a pharmacological activity useful for treating a LBD (e.g., Parkinson's disease or DLBD).
  • the methods involve contacting a transgenic animal expressing alpha-synuclein and processing the alpha-synuclein into a fragment with an agent, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; and determining a level of the fragment in a neuronal cell relative to a baseline level in the absence of the agent, a reduction in the level of the fragments relative to the baseline indicating the agent has the pharmacological activity useful for treating the LBD.
  • a LBD e.g., Parkinson's disease or DLBD
  • the fragment of alpha- synuclein has a C-terminus at a residue between 118 and 125 of intact alpha-synuclein.
  • Preferred fragments include 1-119, 1-120, 1-121, 1-122, 1-123, 1-124, and 1-125 of alpha synuclein.
  • the fragment of alpha-synuclein is 1-X, wherein X is 130-139.
  • the fragment of alpha-synuclein bears a mutation associated with a hereditary LBD, such as an A53T mutation.
  • the transgenic animal is a rodent, mouse or Drosophila.
  • the method involves a step of conducting a trial in a human having a LBD or an animal model of LBD to determine whether the agent treats or inhibits a symptom of the LBD.
  • the invention further provides a transgenic animal having a genome comprising a transgene comprising a promoter operably linked to a nucleic acid segment encoding a fragment of alpha-synuclein wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; wherein expression of the fragment in the transgenic animal disposes the animal to develop at least one characteristic of a LBD.
  • the fragment of alpha-synuclein is selected from the group consisting of 1-119, 1- 120, 1-121, 1-122, 1-123, 1-124, and 1-125.
  • the fragment of alpha-synuclein is 1- X, wherein X is 130-139.
  • the promoter is a PDGF promoter.
  • at least one characteristic is an impairment of motor function.
  • at least one characteristic of the transgenic animal is an impairment of cognitive function.
  • the transgenic animal is a rodent, mouse or Drosophila.
  • the invention further provides methods of detecting presence or susceptibility to an LBD in a patient.
  • the methods involve detecting a level of a fragment of alpha-synuclein in cerebrospinal fluid, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein.
  • a level greater than a baseline level in undiseased individuals indicating presence or susceptibility to LBD.
  • the invention further provides an antibody that specifically binds to a fragment of alpha-synuclein, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; without specifically binding to full-length alpha synuclein.
  • Preferred fragments include 1-119, 1-120, 1-121, 1-122, 1-123, 1-124, and 1-125 of alpha synuclein.
  • the fragment is 1-X, wherein X is 130-139.
  • the antibody is a human, humanized, chimeric antibody.
  • the antibody is monoclonal.
  • the antibody has human isotype IgGl.
  • the invention further provides methods of diagnosing presence or susceptibility to LBD.
  • the methods involve administering to a patient an antibody that specifically binds to a fragment of alpha-synuclein having a free C-terminus at residues 119-125 without specifically binding to full length synuclein; and determining a level of binding of the antibody in the patients, wherein a higher level of binding relative to a base line level in undiseased individuals indicates presence or susceptibility to the LBD.
  • the invention further provides methods of effecting treatment or prophylaxis of a LBD, comprising administering to a patient suffering from or at risk of a LBD, an effective regime of a fragment of alpha-synuclein, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein; and a deletion of at least ten contiguous amino acids from the C-terminus of intact alpha-synuclein, and thereby effecting treatment or prophylaxis of the LBD.
  • the fragment of alpha-synuclein is 1-119, 1-120, 1-121, 1-122, 1-123, 1-124 and 1-125 of alpha synuclein.
  • the fragment is 1-X, wherein X is 130-139.
  • the method further comprises administering an adjuvant that augments an immune response comprising antibodies to the fragment.
  • the fragment is linked to a carrier forming a fusion protein, wherein the carrier augments an immune response comprising antibodies to the fragment.
  • the invention further provides methods of effecting treatment or prophylaxis of a LBD.
  • the method involves administering to a patient suffering from or at risk of a LBD an effective regime of an antibody that specifically binds to a fragment of alpha-synuclein, wherein the fragment is selected from the group consisting of 1-119, 1-120, 1-121, 1-122, 1- 123, 1-124, 1-125 and 1-X, wherein X is 130-139, without binding to intact alpha-synuclein, whereby the antibody effects prophylaxis or treatment of the disease.
  • the invention further provides a method of screening for a protease that cleaves intact alpha-synuclein to form a fragment, wherein the fragment is characterized by presence of at least 100 contiguous amino acids of intact alpha-synuclein and a deletion of 1-23 contiguous amino acids from the C-terminus of intact alpha-synuclein.
  • the method involves identifying an inhibitor of the protease; contacting the inhibitor with a cellular or tissue extract containing the protease, whereby the protease binds to the inhibitor; and releasing the protease from the inhibitor.
  • the inhibitor is a peptide of alpha-synuclein comprising a contiguous segment of at least 5 residues of intact alpha-synuclein between positions 115 and 130.
  • the peptide comprises a contiguous segment of at least 5 residues between positions 118 and 122.
  • at least one of the residues is a transition state analog.
  • the invention further provides a monoclonal antibody that specifically binds to an epitope within residues 109-120 of alpha synuclein.
  • the monoclonal antibody is chimeric, humanized or human.
  • the invention further provides a monoclonal antibody that specifically binds to an epitope within residues 115-123 of alpha synuclein.
  • the invention further provides a monoclonal antibody that specifically binds to a discontinuous epitope within residues 43-51 and 58-65 of alpha synuclein.
  • the antibody is chimeric, humanized or human.
  • the invention further provides an end-specific monoclonal antibody that specifically binds to isolated full-length alpha-synuclein having a free C-terminus without specifically binding to a fusion protein comprising alpha synuclein having a C-terminus linked to a second polypeptide.
  • the antibody is chimeric, humanized or human.
  • the invention further provides methods of detecting presence or susceptibility to a Lewy body disease in a patient.
  • the methods involves determining a level of alpha-synuclein phosphorylated or nitrated at position 125 of alpha-synuclein in a sample from a brain of the patient, an elevated level relative to the mean level in a population of undiseased individuals indicating the patient has or is susceptible to a Lewy body disease.
  • FIGs. 1 A and B show a Western blot of various extracts from the cortex and hippocampus of a transgenic mouse (B) and a matched control (A) with a polyclonal antibody that binds to an epitope within SN115-122.
  • FIG. 2 shows a Western blot with the same antibody as Figs. 1 A and B to compare the level of the truncated form of alpha-synuclein in Triton-XlOO extractions of the cortex and hippocampus mice of 3 months and 12 months in age.
  • Figs. 3A and B shows a Western blot with a different antibody termed 12C1 (a monoclonal binding to epitope at amino acids 43-51 and 58-65) of a Triton extracts from the brain of a transgenic mouse three months old (B) compared with an aged matched control (A).
  • 12C1 a monoclonal binding to epitope at amino acids 43-51 and 58-65
  • Fig. 4 shows a further Western blot using the same antibody as Fig. 3 on a Triton extract from the brain of transgenic mice of three and twelve months of age.
  • FIGs. 5 A, B, C, D, E show Western blots with four different antibodies (B, C, D, E)and an epitope map (A) of the binding sites of the antibodies to various extracts from the brains of transgenic mice.
  • Figs. 6 A, B, C shows Tris extracts of the brain of a patient with Lewy body disease probed with three different antibodies (A, B, C), subject to 2-D gel electrophoresis and subjected to Western blotting.
  • Fig. 7 A, B, C, D shows additional blots of Tris extracts of the brain of a patient with Lewy body disease with four antibodies (A, B, C, D) of additional specificities.
  • Fig. 8 summarizes the sites of cleavage relative to the epitopes bound by antibodies used in the Western blotting.
  • Figs. 9A, B compares the Tris soluble proteins (A) with proteins extracted from Lewy bodies (B) by 2D electrophoresis and Western blotting.
  • Figs. 10 A, B, C, D shows the immunoblots of proteins from Lewy bodies reprobed with various C-terminal antibodies.
  • Figs. 11 shows Western blots of various extracts of an undiseased and Contursi patient probed with an antibody recognizing either total alpha synuclein or specific for phospho-129 alpha synuclein.
  • agent is used to describe a compound that has or may have a pharmacological activity. Agents include compounds that are known drugs, compounds for which pharmacological activity has been identified but which are undergoing further therapeutic evaluation, and compounds that are members of collections and libraries that are to be screened for a pharmacological activity.
  • a "pharmacological" activity means that an agent exhibits an activity in a screening system that indicates that the agent is or may be useful in the prophylaxis or treatment of a disease.
  • the screening system can be in vitro, cellular, animal or human. Agents can be described as having pharmacological activity notwithstanding that further testing may be required to establish actual prophylactic or therapeutic utility in treatment of a disease.
  • the phrases "specifically binds” refers to a binding reaction which is determinative of the presence of the protein in the presence of a heterogeneous population of proteins and other biologies. Thus, under designated conditions, a specified ligand binds preferentially to a particular protein and does not bind in a significant amount to other proteins present in the sample.
  • a molecule such as antibody that specifically binds to a protein often has an association constant of at least 10 6 M " ' or 10 7 M "1 , preferably 10 8 M “1 to 10 9 M “1 , and more preferably, about 10 10 M "1 to 10 11 M “1 or higher.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, e.g., Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat 'I. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally Ausubel et al, supra).
  • HSPs high scoring sequence pairs
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative- scoring residue alignments; or the end of either sequence is reached.
  • the default parameters of the BLAST programs are suitable.
  • the BLASTP program uses as defaults a word length (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix.
  • the TBLATN program (using protein sequence for nucleotide sequence) uses as defaults a word length (W) of 3, an expectation (E) of 10, and a BLOSUM 62 scoring matrix, (see Henikoff & Henikoff, Proc. Natl Acad. Sci. USA 89:10915 (1989)).
  • the BLAST algorithm In addition to calculating percent sequence -identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Nat 1 Acad. Sci. USA 90:5873-5787 (1993)).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • amino acids are grouped as follows: Group I (hydrophobic sidechains): norleucine, met, ala, val, leu, ile; Group II (neutral hydrophilic side chains): cys, ser, thr; Group III (acidic side chains): asp, glu; Group IV (basic side chains): asn, gin, his, lys, arg; Group V (residues influencing chain orientation): gly, pro; and Group VI (aromatic side chains): tip, tyr, phe.
  • Conservative substitutions involve substitutions between amino acids in the same class. Non-conservative substitutions constitute exchanging a member of one of these classes for a member of another.
  • Therapeutic agents of the invention are typically substantially pure from undesired contaminant. This means that an agent is typically at least about 50% w/w (weight/weight) purity, as well as being substantially free from interfering proteins and contaminants. Sometimes the agents are at least about 80% w/w and, more preferably at least 90 or about 95% w/w purity. However, using conventional protein purification techniques, homogeneous peptides of at least 99% w/w can be obtained.
  • antibody or "immunoglobulin” is used to include intact antibodies and binding fragments thereof. Typically, fragments compete with the intact antibody from which they were derived for specific binding to an antigen fragment including separate heavy chains, light chains Fab, Fab' F(ab')2, Fabc, and Fv. Fragments are produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins.
  • antibody also includes one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins.
  • antibody also includes bispecific antibody. A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny et al. , J. Immunol 148, 1547-1553 (1992).
  • adjuvant refers to a compound that when administered in conjunction with an antigen augments the immune response to the antigen, but when administered alone does not generate an immune response to the antigen.
  • adjuvants can augment an immune response by several mechanisms including lymphocyte recruitment, stimulation of B and/or T cells, and stimulation of macrophages.
  • patient includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • competition between antibodies is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as alpha-synuclein.
  • a common antigen such as alpha-synuclein.
  • Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et al, Methods in Enzymology 9:242-253 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et al, J.
  • such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test immunoglobulin and a labeled reference immunoglobulin.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin.
  • the test immunoglobulin is present in excess.
  • Antibodies identified by competition assay include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur.
  • a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50 or 75%.
  • Epitope co-ordinates are approximate ( ⁇ 2 amino acids). Not every amino acid within an epitope is necessarily required for binding.
  • compositions or methods "comprising" one or more recited elements may include other elements not specifically recited.
  • a composition that comprises alpha- synuclein peptide encompasses both an isolated alpha-synuclein peptide and alpha-synuclein peptide as a component of a larger polypeptide sequence.
  • each embodiment, element, step or feature of the invention can be used in combination with any other.
  • the invention is premised in part on the identification of novel fragments of alpha- synuclein in patients with Lewy Body Disease (LBD) and transgenic animal models thereof. These diseases are characterized by aggregations of alpha-synuclein. The fragments have a truncated C-terminus relative to full-length alpha-synuclein. Some fragments are characterized by a molecular weight of about 12 kDa as determined by SDS gel electrophoresis in trycine buffer and a truncation of at least ten contiguous amino acids from the C-terminus of natural alpha-synuclein.
  • the site of cleavage preferably occurs after residue 117 and before residue 126 of natural alpha-synuclein.
  • the identification of these novel fragments of alpha-synuclein has a number of application in for example, drug discovery, diagnostics, therapeutics, and transgenic animals.
  • the invention provides several methods for screening agents for activity useful in treating LBDs. Some methods identify agents that inhibit the cleavage reaction that generates the novel fragments of the invention. Other method identify agents that inhibit aggregation of the products of the cleavage reaction. Such inhibitors are useful for treatment of LBD's. Inhibitors of the cleavage reaction are also useful for affinity purification of the protease responsible for the cleavage reaction.
  • the invention also provides transgenic animal models and cells expressing fragments of alpha-synuclein as described above.
  • the transgenic animal models and cells are disposed to develop characteristics of Lewy body disease, including Lewy bodies containing aggregations of the fragments.
  • the animal models and cells can be used in the screening methods described above.
  • the invention further provides end-specific antibodies that specifically bind to fragments of alpha-synuclein without specifically binding to intact alpha-synuclein per se. These antibodies are useful for in vivo imaging of alpha-synuclein aggregations and also in methods of treatment.
  • the novel alpha-synuclein fragments can also be used in methods of treatment, optionally, in combination with an adjuvant.
  • Alpha-synuclein fragments [0056] Human alpha-synuclein is a peptide of 140 amino acids having the following amino acid sequence:
  • the protein has three recognized domains, a KTKE repeat domain covering amino acids 1-61, a NAC (Non- amyloid component) domain running from about amino acids 60-95, and a C-terminal acidic domain running from about amino acid 98 to 140.
  • Some novel fragments of the invention have C-terminal truncations of at least ten contiguous amino acids, preferably at least 15 contiguous amino acids, and optionally at up to 20, 22, 23 or 25 amino acids.
  • the fragments include all or substantially all (i.e., at least 100 contiguous residues from alpha-synuclein other than the deletion).
  • Some fragments also have relatively short truncations at the N-terminus of up to 20 amino acids, such as deletions of residues 1-4, 1-6, 1-10 and 1-12.
  • Some fragments have N-terminal deletions of residues 1- 23, 1-38 or 1-45.
  • Preferred fragments are SN1-118, SN1-119, SN1-120, SN1-121, SN1-122, SN1-123, SN1-124, SN1-125, SN1-126, SN1-127, SN1-128, SN 1-129 and SN1-130.
  • Particularly preferred fragments are SN1-119, SN1-120, SN1-121, SN1-122, SN1-123, SN 1- 124 and SN 1-125.
  • An especially preferred fragment is SN1-119.
  • the cleavage reaction preferably occurs at a peptide bond between amino acid residues 118 and 126, e.g., between residue 119 and 120.
  • fragments of the invention include N-terminal fragments of alpha-synuclein of about 6 to 7 kDa (as determined by SDS electrophoresis) or 50-80 amino acids.
  • Other fragments of the invention include N-terminal fragments of alpha-synuclein that are free of 1-10 amino acids from the C-terminus of intact alpha-synuclein, i.e., SN 1-X, wherein X is 130-139.
  • Some fragments are characterized by specific binding to antibodies ELADW43 (free N-terminus) and 5C12 (109-120) and lack of specific binding to 8A5 (free C-terminus), LB509 (115-123) and ELAD47 (118-123).
  • Some fragments are characterized by specific binding to ELADW43 (free N-terminus) and 5C12 (109-120), LB509 (115-123) and ELAD47 (118-123) and lack of specific binding to 8A5 (free C-terminus). Some fragments are characterized by specific binding to ELADW43 (free N-terminus) and 5C12 (109-120), LB509 (115-123) and ELAD47 (118-123) and 8A5 (free C-terminus) and lack of specific binding to ELADW43 (free N-terminus).
  • Some fragments or full-length alpha synuclein are phosphorylated or nitrated at the tyrosine residue occupying position 125 of alpha synuclein. Fragments retaining amino acid serine 125 or full-length alpha synuclein can also be phosphorylated at this position. Detection of enhanced phosphorylation or nitration at position 125 or phosphorylation at position 129 in a patient relative to the mean in a population of undiseased individuals is an indication of a Lewy body disease. Detection can be performed using an antibody specific for alpha-synuclein phosphorylated or nitrated at position 125. A level is considered enhanced if greater than the mean plus one standard deviation in a population of undiseased individuals.
  • the fragments of the invention are distinct from the non-A ⁇ component of Alzheimer's disease amyloid (NAC) previously reported. This fragment consisting of at least 28 amino acids residues (residues 60-87) and optionally 35 amino acid residues (residues 61- 95). See Iwai, et al, Biochemistry, 34:10139-10145); Jensen et al, Biochem. J. 310 (Pt 1): 91-94 (1995); GenBank accession number S56746.
  • NAC Alzheimer's disease amyloid
  • alpha-synuclein or its fragments includes the natural human amino acid sequence indicated above, or fragments thereof, as well as analogs including allelic, species and induced variants.
  • Amino acids of analogs are assigned the same numbers as corresponding amino acids in the natural human sequence when the analog and human sequence are maximally aligned.
  • Analogs typically differ from naturally occurring peptides at one, two or a few positions, often by virtue of conservative substitutions.
  • Some natural allelic variants are genetically associated with hereditary LBD. These variants include A30P and A53T.
  • the A53T variation is associated with enhanced levels of phosphorylation at position 129 of alpha synuclein in an individual having the mutation relative to the norm of phosphorylation in undiseased individuals who lack the mutation.
  • Analogs exhibit at least 80 or 90% sequence identity with natural peptides. Some analogs also include unnatural amino acids or modifications of N or C terminal amino acids at one, two or a few positions. For example, the natural glutamic acid residue can be replaced with iso-aspartic acid.
  • unnatural amino acids are D, alpha, alpha-disubstituted amino acids, N-alkyl amino acids, lactic acid, 4-hydroxyproline, gamma-carboxyglutamate, epsilon-N,N,N-trimethyllysine, epsilon-N-acetyllysine, O-phosphoserine, N-acetylserine, N- formylmethionine, 3-methylhistidine, 5-hydroxylysine, omega-N-methylarginine, ⁇ -alanine, ornithine, norleucine, norvaline, hydroxproline, thyroxine, gamma-amino butyric acid, homoserine, citrulline, and isoaspartic acid.
  • Analogs typically specifically bind to a polyclonal antibody population generated against natural human alpha-synuclein.
  • the invention also provides D-peptides, in which D-amino acids can be substituted for corresponding natural L-amino acids of alpha-synuclein at most or all positions.
  • Alpha-synuclein, its fragments, and analogs can be synthesized by solid phase peptide synthesis or recombinant expression, or can be obtained from natural sources. Automatic peptide synthesizers are commercially available from numerous suppliers, such as Applied Biosystems, Foster City, California. Recombinant expression can be in bacteria, such as E. coli, yeast, insect cells or mammalian cells. Procedures for recombinant expression are described by Sambrook et al, Molecular Cloning: A Laboratory Manual (C.S.H.P. Press, NY 2d ed., 1989).
  • Lewy Body Disease is characterized by degeneration of the dopaminergic system, motor alterations, cognitive impairment, and formation of Lewy bodies (LBs).
  • LLB Lewy Body Disease
  • Lewy Bodies are spherical protein deposits found in nerve cells. Their presence in the brain disrupts the brain's normal function interrupting the action of chemical messengers including acetylcholine and dopamine.
  • Lewy Body diseases include Parkinson's disease (including idiopathic Parkinson's disease(PD)), Diffuse Lewy Body Disease (DLBD) also known as Dementia with Lewy Bodies (DLB), Combined Alzheimer's and Parkinson disease and multiple system atrophy (MSA).
  • DLBD differs from Parkinson's disease mainly in the location of Lewy Bodies. In DLBD Lewy Bodies form mainly in the cortex. In Parkinson's disease, they form mainly in the substantia nigra.
  • Lewy Body diseases include Pure Autonomic Failure, Lewy body dysphagia, Incidental LBD, Inherited LBD (e.g., mutations of the alpha-synuclein gene, PARK3 and PARK4) and Multiple System Atrophy (e.g., Olivopontocerebellar Atrophy, Striatonigral Degeneration and Shy-Drager Syndrome).
  • the invention provides transgenic animals having a genome comprising a transgene comprising a nucleic acid segment encoding a C-terminal truncated form of alpha-synuclein as described above.
  • the transgene is preferably present in all or substantially of the somatic and germline cells of the transgenic animal.
  • the nucleic acid segment encoding the C- terminal truncated form of alpha-synuclein is operably linked to one or more regulatory segments that allow the truncated form of alpha-synuclein to be expressed in neuronal cells of the animal.
  • Promoters such as the rat neuron specific enolase promoter, human beta-actin gene promoter, human platelet derived growth factor B (PDGF-B) chain gene promoter, rat sodium channel gene promoter, mouse myelin basic protein gene promoter, human copper- zinc superoxide dismutase gene promoter, and mammalian POU-domain regulatory gene promoter can be used.
  • the PDGF promoter is particularly suitable.
  • an inducible promoter is used.
  • the mouse metallothionine promoter which can be regulated by addition of heavy metals such as zinc to the mouse's water or diet, is suitable.
  • Such transgenic animals can be produced by the same general approaches described by (Masliah et al, Am. J. Pathol.
  • transgenic animals bearing a transgene expressing a truncated alpha-synuclein protein can be crossed with other transgenic models of neurogenic disease, such as models of Alzheimer's disease.
  • transgenic animals bearing a transgene expressing a truncated alpha-synuclein protein can be crossed with transgenic animals bearing a transgene expressed APP with a FAD mutation as described by e.g., Games et al, Nature 373, 523 (1995) McConlogue et al, US 5,612,486, Hsiao et al, Science 274, 99 (1996); Staufenbiel et al, Proc. Natl Acad. Sci. USA 94, 13287-13292 (1997); Sturchler- Pierrat et al, Proc. Natl Acad. Sci.
  • Transgenic animals of the invention are preferably rodents, such as mice or rats, or insects, such as Drosophila.
  • the invention also provides cells transformed with truncated alpha -synuclein which form inclusion bodies containing aggregated truncated alpha-synuclein.
  • the transformed cells are preferably neuronal cells, such as GT1-7 neuronal cells (Hsue et al. Am. J. Pathol. 157:401-410 (2000)), PC12 cells or SY5Y neuroblastoma cells. PEAK cells can also be used.
  • the cells are preferably human cells.
  • a vector comprising a segment encoding a truncated form of alpha-synuclein operably linked to one or more regulatory sequences that ensure expression of the truncated expression is transfected into the cells. Transfected cells can be used to screen agents for activity in clearing alpha-synuclein inclusions.
  • the invention provide several screening methods to identify agents having a pharmacological activity useful in treating a LBD.
  • the methods include screens that can be performed in vitro, in cells or transgenic animals, and which test a variety of parameters as an indication of activity. Agents determined to have an activity in these screens can be retested in secondary screens of animal models of LBD or in clinical trials to determine activity against behavioral or other symptoms of these diseases.
  • the rate or extent of aggregation of alpha-synuclein in the presence of an agent is determined and compared with the rate or extent of aggregation of alpha-synuclein in a contemporaneous or historical control in which the agent was omitted.
  • a reduction in the rate or extent of aggregation in the presence of the agent relative to the control indicates that the agent has activity in inhibiting aggregation of truncated forms of alpha-synuclein. This activity is potentially useful in treating or preventing Lewy Body diseases. 2.
  • Some cellular assays are performed on cells transfected with nucleic acids encoding truncated forms of alpha-synuclein as described above, optionally with a hereditary variation, such as Ala30Pro or Ala53Th. Such cells are contacted with an agent under test, and the rate of extent of aggregation of the truncated alpha-synuclein is measured. The rate of extent of aggregation of alpha-synuclein is then compared to that of similarly transfected control cells in the absence of the agent. Aggregation can be monitored by immunohistochemical analysis, light microscopy or by gel analysis.
  • Gel analysis can detect formation of dimmers, trimers or higher oligomers as well as inability of synuclein to enter gels due to a high level of oligomerization.
  • a reduction in the rate or extent of aggregation in the presence of the test agent relative to the control indicates the agent has activity has a pharmacological activity in inhibiting aggregation of truncated forms of alpha-synuclein. This activity is potentially useful in treating or preventing Lewy Body diseases.
  • Other cellular assays are performed on cells transfected with nucleic acids encoding full-length alpha-synuclein, optionally with a hereditary variation, such as Ala30Pro or Ala53Thr. Such cells are contacted with an agent under test and the rate or extent of formation of truncated forms of alpha-synuclein and/or phosphorylated or nitrated forms of synuclein is/are measured. The presence of these forms can be detected by Western blotting using one or more antibodies to alpha-synuclein. End specific antibodies (i.e., antibodies that bind to a truncated form without binding to full length alpha-synuclein) are particularly useful for this analysis.
  • Collections of antibodies having different epitope specificities can also be used.
  • presence of truncated forms of alpha-synuclein can be shown by presence of bands when blotted with antibodies recognizing an epitope N-terminal of an amino acid segment defined approximately by amino acids 118-125 of intact alpha-synuclein, and , and lack of bands when blotted with an antibody recognizing an epitope C-terminal of this region.
  • the rate or extent of formation of truncated forms of alpha-synuclein and/or phosphorylated or nitrated forms in the presence of agent is compared with that of comparable control cells in the absence of agent.
  • Transgenic animals have a transgene expressing a truncated form of alpha-synuclein as described above, optionally with a hereditary variation, such as Ala30Pro or Ala53Th. Such an animal is contacted with an agent under test, and the rate of extent of aggregation of the truncated form of alpha-synuclein is measured compared with that in a contemporaneous or historical control.
  • the control is usually a similar transgenic animal of the same species that has not been exposed to the agent. Aggregation of alpha-synuclein in a transgenic animal can be monitored by Western blotting or immunohistochemistry as described in the examples.
  • activity of the agent in such transgenic animals can be determined from behavioral characteristics such as motor or cognitive characteristics, as described in the Examples.
  • pharmacological activity of the agent is shown by improved motor or cognitive characteristics (i.e., decrease impairment of such characteristics) relative to a comparable control transgenic animal not exposed to the agent.
  • transgenic animals having a transgene expressing a full-length form of alpha-synuclein, optionally with a hereditary variation, such as Ala30Pro or Ala53Th.
  • a hereditary variation such as Ala30Pro or Ala53Th.
  • Such animals are contacted with an agent under test, and the rate or extent of appearance of truncated forms of alpha-synuclein is detected, optionally with a hereditary variation, such as Ala30Pro or Ala53Th.
  • Such forms can be detected using Western blotting or immunohistochemical analysis using appropriate anti-alpha-synuclein antibodies (as described for the cellular assays).
  • the rate of extent of appearance of truncated forms of alpha-synuclein and/or phosphorylated or nitrated forms is compared with the rate or extent of appearance of such forms in a contemporaneous or historical control constituting a comparable transgenic animal that has not been exposed to the agent.
  • a reduction in the rate or extent of appearance of the truncated forms of alpha-synuclein in the animal exposed to the test agent relative to the control indicates that agent has activity in inhibiting processing of full-length alpha-synuclein to truncated forms.
  • Agents to be screened include antibodies to alpha-synuclein, peptides of alpha- synuclein, drugs known or suspected to have activity in treating a LBD, natural products, and combinatorial libraries.
  • Preferred peptides of alpha-synuclein are relatively short peptides of 30, 25, 20 10 or fewer amino acid including amino acids 118-125 of alpha-synuclein.
  • an amino acid immediately on the N-terminal side of the cleavage site that generates C-terminal truncated forms of alpha-synuclein is replaced with a transition state analog amino acid that forms a nonhydrolizable bond between the two amino acids flanking the cleavage site, e.g., amino acid 119 of alpha synuclein.
  • a transition state analog amino acid that forms a nonhydrolizable bond between the two amino acids flanking the cleavage site, e.g., amino acid 119 of alpha synuclein.
  • transition state analogs are statine, hydroxyethelene, hydroxyethelamine, AHPPA, ACHPA, and derivatives thereof.
  • One or more amino acids of a natural alpha-synuclein sequence can also be substituted with other natural amino acids.
  • Natural products to be screened can also be obtained from the National Cancer Institute's Natural Product Repository, Bethesda, MD. Random libraries of peptides or other compounds can also be screened for suitability. Combinatorial libraries can be produced for many types of compounds that can be synthesized in a step-by-step fashion. Such compounds include polypeptides, beta-turn mimetics, polysaccharides, phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines, oligomeric N-substituted glycines and oligocarbamates.
  • combinatorial libraries of the compounds can be constructed by the encoded synthetic libraries (ESL) method described in Affymax, WO 95/12608, Affymax, WO 93/06121, Columbia University, WO 94/08051, Pharmacopeia, WO 95/35503 and Scripps, WO 95/30642 (each of which is incorporated herein by reference for all purposes).
  • Peptide libraries can also be generated by phage display methods. See, e.g., Devlin, W0 91/18980.
  • Combinatorial libraries and other compounds can initially be screened for suitability by determining their capacity to bind to alpha-synuclein.
  • Analogous strategies to those described in the screening assays can be used to determine whether existing drugs, foods, environmental toxins, and other compounds exert toxic effects via promotion of alpha-synuclein processing, phosphorylation or aggregation. Such assays are performed in the same manner as the screening assays. Toxic activity is indicated by the opposite result to pharmacological activity in the screening assays.
  • protease Processing of full-length alpha-synuclein to the truncated forms of the invention is effected by a protease.
  • the protease can be purified using an inhibitor identified by the screening methods discussed above.
  • a preferred inhibitor is a peptide of alpha-synuclein including residues 117-126 in which a residue N-terminal to the cleavage site has been replaced by a transition state analog.
  • Such an inhibitor is used as an affinity purification reagent to purify the protease from extracts of brain cells.
  • Such cells can be obtained from cadaver of a normal individual or one who has suffered from a LBD disease. Levels of protease may be elevated in the latter.
  • the protease can be assayed by presenting it with an alpha-synuclein substrate and monitoring formation of cleavage products.
  • the substrate can be, for example, the natural human form of alpha-synuclein described above, a fragment thereof containing residues flanking both sides of the cleavage site, or a mutant form thereof in which the mutation is associated with a hereditary form of LBD.
  • the C- terminus of the substrate can be immobilized to the solid phase, and the N-terminus to a label. Cleavage of the substrate releases the label to the liquid phase.
  • the liquid phase can readily be separated from the solid phase, and the amount of label quantified as a measure of proteolytic activity.
  • End-specific antibodies The invention provides end-specific antibodies. Such antibodies specifically bind to a truncated form of alpha-synuclein (at the C-terminus), preferably a form selected of the group consisting of SN1-118, SN1-119, 1-120, 1-121, 1-122, 1-123, 1-124, 1-125 or 1-126 without specifically binding to full-length alpha-synuclein. Such antibodies are useful for in vivo imaging of alpha-synuclein deposits, as therapeutic agents (see below), and for detecting cleavage products resulting from proteolytic cleavage of alpha-synuclein in the screening methods described above.
  • End-specific antibodies are also provided to corresponding C- terminal fragments, e.g., 118-140, 119-140, 120-140, 121-140, 122-140, 123-140, 124-140, 125-140 and 126-140.
  • the end-specific antibodies recognize the N-terminus of these fragments such that they specifically bind to the fragment without specifically binding to full- length alpha synuclein.
  • Such antibodies can be generated by immunizing a laboratory animal with alpha- synuclein or a fragment thereof to induce antibodies, and screening the resulting antibodies to identify those having the desired binding specificity.
  • immunization can be performed with relatively short peptides of less than 20 amino acids that include the C- terminus of the truncated fragments of the invention (e.g., SN 99-118, or SN 110-119).
  • such short peptides are linked to a carrier that helps elicit an immune response.
  • specific binding to a labeled or immobilized truncated fragment can be performed in competition with unlabelled full-length alpha-synuclein.
  • large libraries of antibodies can be screened simultaneously using the phage display technique.
  • non-human monoclonal antibodies e.g., murine, guinea pig, primate, rabbit or rat
  • production of non-human monoclonal antibodies can be performed as described by Harlow & Lane, Antibodies, A Laboratory Manual (CSHP NY, 1988) (incorporated by reference for all purposes).
  • Complete Freund's adjuvant followed by incomplete adjuvant is preferred for immunization of laboratory animals.
  • Rabbits or guinea pigs are typically used for making polyclonal antibodies.
  • Mice are typically used for making monoclonal antibodies. Binding can be assessed, for example, by Western blot or ELISA. The smallest fragment to show specific binding to the antibody defines the epitope of the antibody.
  • epitope specificity can be determined by a competition assay is which a test and reference antibody compete for binding to alpha-synuclein. If the test and reference antibodies compete, then they bind to the same epitope or epitopes sufficiently proximal that binding of one antibody interferes with binding of the other.
  • Chimeric and humanized antibodies have the same or similar binding specificity and affinity as a mouse or other nonhuman antibody that provides the starting material for construction of a chimeric or humanized antibody.
  • Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species.
  • the variable (V) segments of the genes from a mouse monoclonal antibody may be joined to human constant (C) segments, such as IgGl and IgG4.
  • Human isotype IgGl is preferred.
  • the isotype of the antibody is human IgGl .
  • IgM antibodies can also be used in some methods.
  • a typical chimeric antibody is thus a hybrid protein consisting of the V or antigen- binding domain from a mouse antibody and the C or effector domain from a human antibody.
  • Humanized antibodies have variable region framework residues substantially from a human antibody (termed an acceptor antibody) and complementarity determining regions substantially from a mouse-antibody, (referred to as the donor immunoglobulin). See, Queen et al, Proc. Natl. Acad. Sci. USA 86:10029-10033 (1989), WO 90/07861, US 5,693,762, US 5,693,761, US 5,585,089, US 5,530,101, and Winter, US 5,225,539 (each of which is incorporated by reference in its entirety for all purposes).
  • the constant region(s), if present, are also substantially or entirely from a human immunoglobulin.
  • the human variable domains are usually chosen from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine variable region domains from which the CDRs were derived.
  • the heavy and light chain variable region framework residues can be derived from the same or different human antibody sequences.
  • the human antibody sequences can be the sequences of naturally occurring human antibodies or can be consensus sequences of several human antibodies. See Carter et al, WO 92/22653.
  • Certain amino acids from the human variable region framework residues are selected for substitution based on their possible influence on CDR conformation and/or binding to antigen. Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.
  • Human antibodies against alpha-synuclein are provided by a variety of techniques described below. Some human antibodies are selected by competitive binding experiments, or otherwise, to have the same epitope specificity as a particular mouse antibody. Techniques for producing human antibodies include the trioma methodology of Oestberg et al, Hybridoma 2:361-367 (1983); Oestberg, US Patent No.
  • the heavy and light chain variable regions of chimeric, humanized, or human antibodies can be linked to at least a portion of a human constant region.
  • the choice of constant region depends, in part, whether antibody-dependent complement and/or cellular mediated toxicity is desired.
  • isotopes IgGl and IgG3 have complement activity and isotypes IgG2 and IgG4 do not.
  • Choice of isotype can also affect passage of antibody into the brain.
  • Human isotype IgGl is preferred.
  • Light chain constant regions can be lambda or kappa.
  • Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab' F(ab')2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
  • monoclonal antibodies specifically binding to an epitope within residues 109-120, or 115-123, of alpha synuclein, or a discontinuous epitope within residues 43-51 and 68-65, or end-specific to the C-terminus of alpha-synuclein are also provided, including humanized, chimeric and human forms thereof.
  • An end-specific antibody to the C-terminus of alpha-synuclein can be recognized by capacity to specifically bind to alpha-synuclein as a free protein without specifically binding to alpha synuclein as a component of a fusion protein when the C-terminus of alpha-synuclein is linked to a second peptide.
  • the invention provides methods of in vivo imaging LBs in a patient. Such methods are useful to diagnose or confirm diagnosis of a Lewy Body disease of PD or susceptibility thereto. For example, the methods can be used on a patient presenting with symptoms of dementia. If the patient has LBs, then the patient is likely suffering from a Lewy Body disease. The methods can also be used on asymptomatic patients. Presence of abnormal deposits of amyloid indicates susceptibility to future symptomatic disease. The methods are also useful for monitoring disease progression and/or response to treatment in patients who have been previously diagnosed with a Lewy Body disease.
  • the methods work by administering an end-specific antibody as described above that binds to alpha-synuclein in the patient and then detecting the antibody after it has bound. If desired, a clearing response can be avoided by using antibody fragments lacking a full length constant region, such as Fabs. In some methods, the same antibody can serve as both a treatment and diagnostic reagent.
  • Diagnostic reagents can be administered by intravenous injection into the body of the patient, or directly into the brain by intracranial injection or by drilling a hole through the skull.
  • the dosage of reagent should be within the same ranges as for treatment methods.
  • the reagent is labeled, although in some methods, the primary reagent with affinity for alpha-synuclein is unlabelled and a secondary labeling agent is used to bind to the primary reagent.
  • the choice of label depends on the means of detection. For example, a fluorescent label is suitable for optical detection. Use of paramagnetic labels is suitable for tomographic detection without surgical intervention. Radioactive labels can also be detected using PET or SPECT.
  • Diagnosis is performed by comparing the number, size and/or intensity of labeled loci to corresponding base line values.
  • the base line values can represent the mean levels in a population of undiseased individuals. Base line values can also represent previous levels determined in the same patient. For example, base line values can be determined in a patient before beginning treatment, and measured values thereafter compared with the base line values. A decrease in values relative to base line signals a positive response to treatment.
  • End-specific antibodies are also useful to determine whether truncated forms of alpha-synuclein are present in cerebrospinal fluid or other body tissues or fluids. Presence of such forms at significantly greater levels (i.e., greater than mean plus one standard deviation) in a patient relative to the normal level in a population of undiseased individuals is indicative of presence or susceptibility to a LBD.
  • the invention provides several methods of preventing or treating Lewy Body disease in patients suffering from or at risk of such disease.
  • Therapeutic agents include the truncated forms of alpha-synuclein described above, and fragments thereof effective to induce antibodies, end-specific antibodies as described above, and inhibitors of aggregation of truncated fragments of alpha-synuclein or proteolytic processing of alpha-synuclein as described above.
  • Patients amenable to treatment include individuals at risk of disease of a LBD but not showing symptoms, as well as patients presently showing symptoms. Therefore, the present methods can be administered prophylactically to individuals who have a known genetic risk of a LBD. Such individuals include those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers. Genetic markers of risk toward PD include mutations in the alpha-synuclein or Parkin, UCHLI, and CYP2D6 genes; particularly mutations at positions 30 and 53 of the alpha-synuclein gene. Individuals presently suffering from Parkinson's disease can be recognized from its clinical manifestations including resting tremor, muscular rigidity, bradykinesia and postural instability.
  • the patient is free of clinical symptoms or risk factors any amyloidogenic disease other than one characterized by Lewy bodies. In some methods, the patient is free of clinical symptoms or risk factors of any disease characterized by extracellular amyloid deposits. In some methods, the patient is free of diseases characterized by amyloid deposits of AjS peptide. In some methods, the patient is free of clinical symptoms and risk factors of Alzheimer's disease. In some methods, the patient has concurrent Alzheimer's disease and a disease characterized by Lewy bodies. In some methods, the patient has concurrent Alzheimer's and Parkinson's disease.
  • treatment can begin at any age (e.g., 10, 20, 30). Usually, however, it is not necessary to begin treatment until a patient reaches 40, 50, 60 or 70. Treatment typically entails multiple dosages over a period of time. Treatment can be monitored by assaying antibody, or activated T-cell or B-cell responses to a therapeutic agent (e.g., a truncated form of alpha-synuclein peptide) over time. If the response falls, a booster dosage is indicated.
  • a therapeutic agent e.g., a truncated form of alpha-synuclein peptide
  • compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of a LBD in regime comprising an amount and frequency of administration of the composition or medicament sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including physiological, biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • compositions or medicates are administered to a patient suspected of, or already suffering from such a disease in a regime comprising an amount and frequency of administration of the composition sufficient to cure, or at least partially arrest, the symptoms of the disease (physiological, biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease.
  • An amount adequate to accomplish therapeutic or prophylactic treatment is defined as a therapeutically- or prophylactically-effective dose.
  • a combination of amount and dosage frequency adequate to accomplish therapeutic or prophylactic treatment is defined as a therapeutically or prophylatically-effective regime.
  • agents are usually administered in several dosages until a sufficient immune response has been achieved. Typically, the immune response is monitored and repeated dosages are given if the immune response starts to wane.
  • administration of an agent results in reduction of intracellular levels of aggregated alpha-synuclein. In some methods, administration of the agent results in a reduction in levels of C-terminal truncated forms of alpha-synculein. In some methods, administration of an agent results in improvement in a clinical symptom of a LBD, such as motor or cognitive function in the case of Parkinson's disease. In some methods, reduction in intracellular levels of aggregated alpha-synuclein or improvement in a clinical symptom of disease is monitored at intervals after administration of an agent.
  • Effective doses of the compositions of the present invention, for the treatment of the above described conditions vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic.
  • the patient is a human but nonhuman mammals including transgenic mammals can also be treated. Treatment dosages need to be titrated to optimize safety and efficacy.
  • the agent is a truncated fragment of alpha-synuclein or a fragment thereof capable of inducing antibodies to alpha-synuclein.
  • the amount of such a fragment depends on whether adjuvant is also administered, with higher dosages being required in the absence of adjuvant.
  • the amount of a fragment for administration sometimes varies from 1- 500 ⁇ g per patient and more usually from 5-500 ⁇ g per injection for human administration. Occasionally, a higher dose of 1-2 mg per injection is used. Typically about 10, 20, 50 or 100 ⁇ g is used for each human injection.
  • the mass of fragment also depends on the mass ratio of immunogenic epitope within the fragment to the mass of fragment as a whole.
  • 10 "3 to 10 "5 micromoles of immunogenic epitope are used for microgram of fragment.
  • the timing of injections can vary significantly from once a day, to once a year, to once a decade. On any given day that a dosage of immunogen is given, the dosage is greater than 1 ⁇ g/patient and usually greater than 10 ⁇ g/ patient if adjuvant is also administered, and greater than 10 ⁇ g/patient and usually greater than 100 ⁇ g/patient in the absence of adjuvant.
  • a typical regimen consists of an immunization followed by booster injections at time intervals, such as 6 week intervals. Another regimen consists of an immunization followed by booster injections 1, 2 and 12 months later. Another regimen entails an injection every two months for life. Alternatively, booster injections can be on an irregular basis as indicated by monitoring of immune response.
  • Truncated fragments of alpha-synuclein can also be administered in the form of nucleic acids encoding the fragments operably linked to one or more regulatory elements to ensure expression of the truncated fragment of alpha-synuclein.
  • Doses for nucleic acids encoding immunogens range from about 10 ng to 1 g, 100 ng to 100 mg, 1 ⁇ g to 10 mg, or 30-300 ⁇ g DNA per patient.
  • Doses for infectious viral vectors vary from 10-100, or more, virions per dose.
  • Some methods involve passive immunization with an end-specific antibody.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be 1 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg or, in other words, 70 mg or 700 mg or within the range of 70-700 mg, respectively, for a 70 kg patient.
  • An exemplary treatment regime entails administration once per every two weeks or once a month or once every 3 to 6 months.
  • two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody administered falls within the ranges indicated.
  • Antibody is usually administered on multiple occasions.
  • Intervals between single dosages can be weekly, monthly or yearly. Intervals can also be irregular as indicated by measuring blood levels of antibody to alpha-synuclein in the patient. In some methods, dosage is adjusted to achieve a plasma antibody concentration of 1-1000 ⁇ g/ml and in some methods 25-300 ⁇ g/ml. Alternatively, antibody can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic.
  • a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives.
  • a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patent can be administered a prophylactic regime.
  • Therapeutic agents can be administered by parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intracranial, intrathecal, intraperitoneal, intranasal or intramuscular means for prophylactic and/or therapeutic treatment.
  • the most typical route of administration of an immunogenic agent is subcutaneous although other routes can be equally effective.
  • the next most common route is intramuscular injection. This type of injection is most typically performed in the arm or leg muscles.
  • agents are injected directly into a particular tissue where deposits have accumulated, for example intracranial injection, intramuscular injection or intravenous infusion is preferred for administration of antibody.
  • particular therapeutic antibodies are injected directly into the cranium.
  • antibodies are administered as a sustained release composition or device, such as a MedipadTM device.
  • Small molecules that act by inhibiting protease processing of alpha-synuclein can be administered intravenously if the small molecules pass through the blood brain barrier sufficiently for therapeutic or prophylactic efficacy or directly into the cranium otherwise.
  • Agents of the invention can optionally be administered in combination with other agents that are at least partly effective in treatment of LBD. Agents of the invention can also be administered in conjunction with other agents that increase passage of the agents of the invention across the blood-brain barrier.
  • Immunogenic agents are sometimes administered in combination with an adjuvant.
  • adjuvants can be used in combination with a peptide, such as alpha-synuclein, to elicit an immune response.
  • Preferred adjuvants augment the intrinsic response to an immunogen without causing conformational changes in the immunogen that affect the qualitative form of the response.
  • Preferred adjuvants include aluminum hydroxide and aluminum phosphate, 3 De-O-acylated monophosphoryl lipid A (MPLTM) (see GB 2220211 (RIBI ImmunoChem Research Inc., Hamilton, Montana, now part of Corixa).
  • Stimulon 1 M QS-21 is a trite ⁇ ene glycoside or saponin isolated from the bark of the Quillaja Saponaria Molina tree found in South America (see Kensil et al, in Vaccine Design: The Subunit and Adjuvant Approach (eds. Powell & Newman, Plenum Press, NY, 1995); US Patent No. 5,057,540), (Aquila BioPharmaceuticals, Framingham, MA).
  • Other adjuvants are oil in water emulsions (such as squalene or peanut oil), optionally in combination with immune stimulants, such as monophosphoryl lipid A (see Stoute et al, N. Engl J Med.
  • Adjuvants can be administered as a component of a therapeutic composition with an active agent or can be administered separately, before, concurrently with, or after administration of the therapeutic agent.
  • a preferred class of adjuvants is aluminum salts (alum), such as alum hydroxide, alum phosphate, alum sulfate. Such adjuvants can be used with or without other specific immunostimulating agents such as MPL or 3-DMP, QS-21, polymeric or monomeric amino acids such as polyglutamic acid or polylysine.
  • Another class of adjuvants is oil-in-water emulsion formulations.
  • Such adjuvants can be used with or without other specific immunostimulating agents such as muramyl peptides (e.g., N-acetylmuramyl-L-threonyl-D- isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N- acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-( 1 '-2'dipalmitoyl-sn-glycero-3 - hydroxyphosphoryloxy)-ethylamine (MTP-PE), N-acetylglucsaminyl-N-acetylmuramyl-L- Al-D-isoglu-L-Ala-dipalmitoxy propylamide (DTP-DPP) theramideTM), or other bacterial cell wall components.
  • Oil-in-water emulsions include (a) MF59 (WO 90/14837), containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE) formulated into submicron particles using a micro fluidizer such as Model 110Y micro fluidizer (Micro fluidics, Newton MA), (b) SAF, containing 10% Squalene, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi ImmunoChem, Hamilton, MT) containing 2% squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylhpid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton
  • Another class of preferred adjuvants is saponin adjuvants, such as StimulonTM (QS- 21, Aquila, Framingham, MA) or particles generated therefrom such as ISCOMs (immunostimulating complexes) and ISCOMATRIX.
  • Other adjuvants include RC-529, GM- CSF and Complete Freund's Adjuvant (CFA) and Incomplete Freund's Adjuvant (IF A).
  • adjuvants include cytokines, such as interleukins (e.g., IL-1, IL-2, IL-4, IL-6, IL-12, IL13, and IL-15), macrophage colony stimulating factor (M-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), and tumor necrosis factor (TNF).
  • cytokines such as interleukins (e.g., IL-1, IL-2, IL-4, IL-6, IL-12, IL13, and IL-15), macrophage colony stimulating factor (M-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), and tumor necrosis factor (TNF).
  • M-CSF macrophage colony stimulating factor
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • Another class of adjuvants is glycolipid analogues including N-glycosylamides, N
  • An adjuvant can be administered with an alpha-synuclein fragment as a single composition, or can be administered before, concurrent with or after administration of the alpha-synuclein fragment .
  • the alpha-synuclein fragment and adjuvant can be packaged and supplied in the same vial or can be packaged in separate vials and mixed before use.
  • the alpha-synuclein fragment and adjuvant are typically packaged with a label indicating the intended therapeutic application. If the alpha-synuclein fragment and adjuvant are packaged separately, the packaging typically includes instructions for mixing before use.
  • an adjuvant and/or carrier depends on the stability of the immunogenic formulation containing the adjuvant, the route of administration, the dosing schedule, the efficacy of the adjuvant for the species being vaccinated, and, in humans, a pharmaceutically acceptable adjuvant is one that has been approved or is approvable for human administration by pertinent regulatory bodies.
  • Complete Freund's adjuvant is not suitable for human administration.
  • Alum, MPL and QS-21 are preferred.
  • two or more different adjuvants can be used simultaneously. Preferred combinations include alum with MPL, alum with QS-21, MPL with QS-21, MPL or RC-529 with GM-CSF, and alum, QS-21 and MPL together.
  • incomplete Freund's adjuvant can be used (Chang et al, Advanced Drug Delivery Reviews 32, 173-186 (1998)), optionally in combination with any of alum, QS-21, and MPL and all combinations thereof.
  • Agents of the invention are often administered as pharmaceutical compositions comprising an active therapeutic agent, i.e., and a variety of other pharmaceutically acceptable components. See Remington's Pharmaceutical Science (15th ed., Mack Publishing Company, Easton, Pennsylvania, 1980). The preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination.
  • compositions or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.
  • compositions can also include large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized Sepharose(TM), agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes). Additionally, these carriers can function as immunostimulating agents (i.e., adjuvants).
  • agents of the invention can be administered as injectable dosages of a solution or suspension of the substance in a physiologically acceptable diluent with a pharmaceutical carrier that can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
  • a pharmaceutical carrier that can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
  • auxiliary substances such as wetting or emulsifying agents, surfactants, pH buffering substances and the like can be present in compositions.
  • Other components of pharmaceutical compositions are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, and mineral oil.
  • glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
  • Antibodies can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained release of the active ingredient.
  • An exemplary composition comprises monoclonal antibody at 5 mg/mL, formulated in aqueous buffer consisting of 50-mM L-histidine, 150 mM NaCl, adjusted to pH 6.0 with HC1.
  • Compositions for parenteral administration are typically substantially sterile, substantially isotonic and manufactured under GMP conditions of the FDA or similar body.
  • compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above (see Langer, Science 249, 1527 (1990) and Hanes, Advanced Drug Delivery Reviews 28, 97-119 (1997).
  • the agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.
  • Additional formulations suitable for other modes of administration include oral, intranasal, and pulmonary formulations, suppositories, and transdermal applications.
  • binders and carriers include, for example, polyalkylene glycols or triglycerides; such suppositories can be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably l%-2%.
  • Oral formulations include excipients, such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, and magnesium carbonate. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10%-95% of active ingredient, preferably 25%-70%.
  • Topical application can result in transdermal or intradermal delivery.
  • Topical administration can be facilitated by co-administration of the agent with cholera toxin or detoxified derivatives or subunits thereof or other similar bacterial toxins (See Glenn et al., Nature 391, 851 (1998)).
  • Co-administration can be achieved by using the components as a mixture or as linked molecules obtained by chemical crosslinking or expression as a fusion protein.
  • transdermal delivery can be achieved using a skin path or using transferosomes (Paul et al, Eur. J. Immunol. 25, 3521-24 (1995); Cevc et al, Biochem. Biophys. Ada 1368, 201-15 (1998)).
  • Examples 1 Detecting truncated forms of alpha-synuclein in a transgenic animal [0111] Transgenic mice having a nucleic acid encoding intact alpha-synuclein operably linked to a PDFG promoter were analyzed at 6 weeks, 3 months and 12 months old. The animals were euthanized and the cortex and hippocampus tissue from four mice (2 male/2 female) was pooled. The tissue was homogenized in TBS (250 mM NaCl), and spun at 150,000 x g for 15 minutes. The pellet was then extracted with 1% Triton-X 100 for 30 min at 4 degrees and spun as before. The resulting pellet was then extracted with 1% SDS for 30 min at 25 degrees and spun as before. Finally, the pellet was extracted with 8 M Urea/ 1% SDS. This procedure resulted in four extracts which will be referred to as Tris, Triton, SDS, and Urea extracts in the description that follows.
  • Figs. 1 A and B show a Western blot of extracts from a transgenic mouse and a matched control with antibody ELADW-47.
  • This antibody is a polyclonal that binds to an epitope within SN115-122 (but does not necessarily require each amino acid for some binding to occur).
  • the antibody preferentially binds the human form of alpha-synuclein but also binds the mouse form to a lesser extent.
  • Figs. 1 A and B shows an alpha-synuclein band at 14 kDa for both the control mouse and the transgenic mouse. The band is stronger for the transgenic mouse than the control. For the different extracts, the band is most intense in the Triton extract.
  • Tris and particularly the Triton extractions of the transgenic mouse show a band at about 12 kDa in a trycine buffer. This is a truncated form of alpha-synuclein.
  • the molecular weight of the band corresponds to a length of about 115-120 amino acids.
  • Fig. 2 shows a Western blot with the same antibody as Fig. 1 to compare the level of the truncated form of alpha-synuclein in mice of 3 months and 12 months in age.
  • the Figure shows that the truncated form appears more strongly in the 3 month old mice. Again, the truncated band does not appear in the control mice.
  • the more intense appearance of the truncated form of alpha-synuclein early in development of the transgenic mice indicates that the truncated form of alpha-synuclein has a role early in the pathogenesis of Lewy Body disease.
  • Figs. 3A and B show a Western blot with a different antibody termed 12C1 (binds epitope at amino acids 43-51 and 58-65, monoclonal, IgGl k).
  • This antibody binds equally to mouse and human forms of alpha-synuclein at an epitope including amino acids 43-51 and 58-65.
  • Fig. 3 shows the truncated band of 12 kDa in the Triton extract of the transgenic mice. The same band appears much more faintly in the Triton extract of the control mice.
  • processing of alpha-synuclein to a truncated form occurs in both normal mice and transgenic mice, but more strongly in the latter.
  • the greater extent of processing in the transgenic mice may be due to processing of the human alpha-synuclein directly, or may be due to the presence of human alpha-synuclein driving mouse alpha-synuclein down a path that is used to a lesser extent in nontransgenic mice.
  • Fig. 4 shows a further Western blot using the same antibody as Fig. 3.
  • This gel shows two additional bands of molecular weights approximately 6 or 7 kDa.
  • the 7 kDa band appears more strongly in the transgenic mice than control mice.
  • the 6 kDa band appears only in the transgenic mouse, and then only in the 3 mo sample.
  • the 6 or 7 kDa bands are indicative of shorter N-terminal fragments of alpha-synuclein of length about 50-80 amino acids.
  • Figs. 5 A, B, C, D, E shows Western blots with four different antibodies and epitope maps of the binding sites of the antibodies.
  • ELADW-44 is a polyclonal that binds only to the human form of alpha-synuclein (i.e., not the mouse form) It binds to at epitope at amino acids 98-019.
  • ELADW-47 is a polyclonal that binds preferentially to the human form but also binds the mouse form. It binds to an epitope at amino acids 115-122.
  • ELADW-48 is a polyclonal that binds the human and mouse forms equally. It binds to an epitope between amino acids 131 and 140.
  • 8A5 is a monoclonal that binds to the human and mouse forms equally. It binds to the C-terminus of alpha-synuclein.
  • Figs. 5A-E shows that of these four antibodies, only ELADW-47 generated a 12 kDa band indicative of a truncated form of alpha-synuclein. The result that ELADW48 did not give rise to this band is of assistance in mapping the site of cleavage. Because ELAD-47 did bind and ELAD-48 did not the site of cleavage is bordered by the N-terminal end of the ELADW-47 epitope and the C-terminal amino acid of the ELADW48 epitope.
  • the cleavage site is further confined to a region approximately within amino acids 118-135.
  • the probable site of cleavage is around amino acids 118-121.
  • the lack of binding by the C-terminal antibody 8A5 is consistent with this cleavage site.
  • the lack of binding by the antibody ELADW-44 requires further comment.
  • cleavage can be explained if a truncated form of human alpha- synuclein resulting from cleavage adapts a different conformation to intact alpha-synuclein preventing biding of ELDAW-44.
  • the truncated form of alpha-synuclein present in transgenic mice to a greater extent than in normal mice represent a form of mouse alpha-synuclein.
  • the greater amount of the truncated form in the transgenic mouse would be due to the presence of the human alpha-synuclein driving more of the mouse alpha-synuclein down a processing path that leads to truncated alpha-synuclein relative to the situation in a control mouse.
  • LBs and soluble protein were prepared from the cortex of a single DLBD patient (see Jensen et al., J. Biol. Chem. 275 21500-21507 (2000)). Tissue was homogenized in Tris/sucrose (0.32 mM)/EDTA (5mM) and protease inhibitors buffer. The homogenate was spun at lOOOg. The supernatant was subjected to a further spin at 150 kg. The supernatant from this spin was used to prepare a Tris soluble fraction of proteins.
  • the pellet from the lOOOg was resuspended and used to prepare a Lewy body fraction.
  • Lewy bodies were purified by immunoprecipitation on magnetic beads bearing anti-synuclein antibodies.
  • the precipitate was then extracted with 7 M Urea 2 M Thiourea/4% CHAPS.
  • the bound form was reextracted with Urea/Thiourea/CHAPS.
  • the extracts from this step and the previous extraction were then pooled and analyzed by 2D PAGE and immunoblot.
  • the bound form was subject to further extraction with 90% formic acid.
  • the extract was stored diluted to 9% formic acid.
  • the extract was then analyzed by SDS PAGE and RP-HPLC.
  • alpha-synuclein species including phosphorylated and truncated species, were present in both LBs and the soluble brain fraction.
  • the predominant truncations were in the C-terminal region of alpha-synuclein at approximately amino acids 120-125.
  • An additional larger fragment cleaved close to the C-terminus was also observed.
  • No beta or gamma- synuclein was detected in the LBs despite being found in the soluble protein fraction.
  • the alpha-synuclein in the LB preparation differed from that in the soluble fraction in that it had additional C-terminal cleavages, and that overall the truncated alpha-synuclein species were enriched in the LBs relative to the soluble protein fraction.
  • multiple alpha- synuclein species of higher molecular mass approximately 25-35 kDa, were detected only in the LB preparation.
  • the C-terminally truncated fragments are of the same size as those observed in the transgenic mouse model of Example 1 indicating a role in disease pathogenesis.
  • Figs. 6A, B, C shows Tris extracts probed with different antibodies, subject to 2-D gel electrophoresis and subjected to Western blotting.
  • the dark spots present toward the left of the charts represent full-length alpha-synuclein.
  • the most notable feature is four spots in the Syn-1 chart that are absent in the 8A5 chart. These four spots represent truncated forms of alpha-synuclein that are unable to bind the 8A5 antibody because of the lack of a C- terminal amino acid. These truncations correspond approximately to forms of SN between 1- 120 and 1-125. Several additional spots are seen underneath and adjacent to the full length alpha-synuclein spots.
  • the spots underneath the full length spots probably represent minor truncations from the C-terminus (i.e., synuclein 1-X, wherein X is 130-139).
  • the spot adjacent the full-length spots but to the right represent a minor deletion from the N-terminus (due to lack of this spot in the blot with ELADW43).
  • Figs. 7 A, B, C, D shows blots with additional antibodies.
  • the four spots are present with 5C12 (109-120).
  • Two of the spots are present with ELADW47 (118-123) and the spots are absent with LB509 (115-123).
  • the spots may differ from each other both in molecular weight and in the presence or absence of posttranslational modification, such as nitration or phosphorylation. These results fix the sites of cleavage to within about amino acids 120-125 of alpha-synuclein.
  • spots running slight below (lower molecular weight) or to the right (higher pH) than the unmodified synuclein spots These likely may represent forms of synuclein that have undergone a small extent of truncation and/or different posttranslational modification relative to the main spots.
  • Fig. 8 summarizes the sites of cleavage relative to the epitopes bound by antibodies used in the Western blotting.
  • Figs. 9 A, B compare the Tris soluble proteins with proteins extracted from Lewy bodies by 2D electrophoresis and Western blotting.
  • the Tris blot on the left shows four spots at lower molecular weight representing truncated forms of alpha-synuclein (probably in the 1- 120 5o 1-125 amino acid range). These are of relatively low intensity compared to the spots representative of full length alpha-synuclein.
  • the blot of proteins from Lewy bodies shows more spots representative of truncated forms of alpha synuclein in the 1-120 to 1-125 range. However, these are of greater intensity relative to the spots representative of full length alpha synuclein.
  • Figs. 10A, B, C, D show the immunoblots of proteins from Lewy bodies reprobed with various C-terminal antibodies. All spots appear with Syn-1 (91-96) and 5C12 (109- 120). With ELADW47, the spot running at the fastest speed and most basic position in the Syn-1 and 5C12 blots is missing. In the LB509 blot, all of the faster moving/more basis spots in the other blots are missing or faint. The absence or reduced intensity of certain spots in the ELADW47 and LB509 blots indicates that these spots represent truncated forms of alpha- synuclein and are consistent with cleavage occurring approximately between amino acids 120 and 125. 3. Detecting Aggregated Alpha-Synuclein in a Transgenic Animal
  • Transgenic animals are euthanized and brains are removed for neurochemical and neuropathological analysis Briefly, the right hemibrain is frozen and homogenized for determinations of aggregated and non-aggregated human alpha-synuclein immunoreactivity by Western blot (Masliah et al, Science (2000) 287:1265). The left hemibrain is fixed in 4% paraformaldehyde, serially sectioned in the vibratome for immunocytochemistry and ultrastructural analysis.
  • Brain sections are immunostained with a rabbit polyclonal antibody against human alpha-synuclein (1:500). After an overnight incubation at 4°C, sections are incubated with biotinylated anti-rabbit secondary antibody followed by Avidin D-Horseradish peroxidase (HRP) complex (1 :200, ABC Elite, Vector). The reaction is visualized with 0.1% 3,3,- diaminobenzidine tetrahydrochloride (DAB) in 50mM Tris-HCl (pH 7.4) with 0.001% H 2 O 2 and sections are then mounted on slides under Entellan. Levels of immunoreactivity are semi quantitatively assessed by optical densitometry using the Quantimet 570C.
  • HRP Avidin D-Horseradish peroxidase
  • Sections will be imaged with the LSCM and each individual image is interactively thresholded such that the TH-immunoreactive terminals displaying pixel intensity within a linear range are included.
  • a scale is set to determine the pixel to ⁇ m ratio. Then, this information is used to calculate the % area of the neuropil covered by TH-immunoractive terminals.
  • MSA multiple system atrophy
  • A53T familial Parkinson's disease mutation
  • Particulate fractions of MSA and Contursi brain were prepared by homogenizing brain tissue in 50 mM Tris, 140 mM NaCl and either l%Triton (MSA) or 0.1% NP40 (Contursi) respectively
  • MSA l%Triton
  • NP40 contursi
  • the insolubility of synuclein in the Contursi brain was associated with a large increase in synuclein phosphorylation at ser 129.
  • the ⁇ -synuclein in the Contursi patient also differed from that in normal brain in the distribution of C-terminal truncations. C-terminally truncated ⁇ -synuclein were observed in both control and Contursi particulate brain fraction
  • all detectable truncations were highly insoluble (urea/SDS extract) in the Contursi patient, whereas those in the control brain were soluble (tris buffered sucrose extract).
  • the enrichment of the C-terminally truncated synuclein in a LB-enriched fraction of a Contursi patient is in agreement with our finding of C-terminally truncated synuclein enrichment in DLBD LBs.
  • the MSA brain was also enriched in phospho (ser 129)- ⁇ -synuclein revealed C-terminal truncation and an abundance of phosphorylation and other acidic modifications also seen in LBs. High levels of phospho (ser 129) were also seen in the brain of an DLBD patient relative to an undiseased control.
  • mice are analyzed for 2 days in the rotarod (San Diego) Instruments, San Diego, CA), as previously described (Masliah, et al. (2000)). On the first day mice are trained for 5 trials: the first one at 10 ⁇ m, the second at 20 ⁇ m and the third to fifth at 40 ⁇ m. On the second day, mice are tested for 7 trials at 40 ⁇ m each. Mice are placed individually on the cylinder and the speed of rotation is increased from 0 to 40 ⁇ m over a period of 240 sec. The length of time mice remain on the rod (fall Latency) is recorded and used as a measure of motor function.
  • mice are tested for cognitive ability in the Morris Water maze (Morris, Learn Motivat. 12;239-260 (1981)).
  • the animal is placed in a circular pool filled with water, with an escape platform submerged just below the surface of the water.
  • a visible marker is placed on the platform so that the animal can find it by navigating toward a proximal visual cue.
  • a more complex form of the test in which there are no formal cues to mark the platform's location will be given to the animals. In this form, the animal must learn the platform's location relative to distal visual cues. The length of time the animal remains in the water is inversely related to its cognitive ability.
  • GT1-7 neuronal cells are transfected with a pCR3.1-T expression vector (Invitrogen, Carlsbad, CA) expressing a truncated fragment of alpha-synuclein as described above murine alpha-synuclein and compared with cells transfected with expression vector alone
  • a pCR3.1-T expression vector Invitrogen, Carlsbad, CA
  • Cells transfected with vector alone have a fibroblastic appearance while cells transfected with alpha-synuclein are rounded, with inclusion bodies at the cell surface visible via both light and confocal scanning microscopy.
  • Transfected GT1-7 cells can be used to screen agents for activity in clearing synuclein inclusions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Neurology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Plant Pathology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Environmental Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Husbandry (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Psychology (AREA)

Abstract

La présente invention concerne de nouveaux fragments d'α-synucléine chez des patients atteints de maladie affectant les corps de Lewy, et des modèles animaux transgéniques de ces fragments. Ces maladies sont caractérisées par une aggrégation d'α-synucléine. Ces fragments présentent une terminaison C tronquée par rapport à l'α-synucléine complète. Certains fragments sont caractérisés, d'une part par une masse moléculaire d'environ 12 kDa, mesurée par électrophorèse sur gel SDS dans un tampon de trycine, et d'autre part par une troncation d'au moins dix acides aminés à partir de la terminaison C de l'α-synucléine naturelle. Le site de clivage se présente de préférence après le résidu 117 et avant le résidu 126 de l'α-synucléine naturelle. L'identification de ces nouveaux fragments d'α-synucléine trouve plusieurs applications dans les domaines, notamment, de la découverte de médicaments, des diagnostics, des thérapies, et des animaux trasngéniques.
PCT/US2004/015836 2003-05-19 2004-05-19 Fragments tronques d'alpha-synucleine dans les affections des corps de lewy WO2005013889A2 (fr)

Priority Applications (14)

Application Number Priority Date Filing Date Title
EP04776059.0A EP1633189B1 (fr) 2003-05-19 2004-05-19 Fragments tronques d'alpha-synucleine dans les affections des corps de lewy
DK04776059.0T DK1633189T3 (en) 2003-05-19 2004-05-19 TRUNCTED FRAGMENTS OF ALFA SYNUCLEIN IN LEWY BODY DISEASE
SI200432403T SI1633189T1 (sl) 2003-05-19 2004-05-19 Skrajšani fragmenti alfa-sinukleina pri bolezni Lewyjevih telesc
ES04776059.0T ES2640669T3 (es) 2003-05-19 2004-05-19 Fragmentos truncados de alfa-sinucleína en enfermedad con cuerpos de lewy
CA2526900A CA2526900C (fr) 2003-05-19 2004-05-19 Fragments tronques d'alpha-synucleine dans les affections des corps de lewy
JP2006514909A JP4820291B2 (ja) 2003-05-19 2004-05-19 レヴィー小体病におけるαシヌクレインの切断断片
PL04776059T PL1633189T3 (pl) 2003-05-19 2004-05-19 Skrócone fragmenty alfa-synukleiny w chorobie z ciałami lewy'ego
US10/969,335 US7306945B2 (en) 2003-05-19 2004-10-19 Truncated fragments of alpha-synuclein in Lewy body disease
US11/194,115 US7358331B2 (en) 2003-05-19 2005-07-29 Truncated fragments of alpha-synuclein in Lewy body disease
US12/040,739 US20090035217A1 (en) 2003-05-19 2008-02-29 Truncated Fragments of Alpha-Synuclein in Lewy Body Disease
US13/604,441 US9879071B2 (en) 2003-05-19 2012-09-05 Antibodies to alpha synuclein
US15/466,543 US20170204173A1 (en) 2003-05-19 2017-03-22 Antibodies to Alpha Synuclein
CY20171100989T CY1119556T1 (el) 2003-05-19 2017-09-20 Περικομμενα θραυσματα της αλφα-συνουκλεϊνης σε νοσο με σωματια lewy
US15/981,008 US20180355028A1 (en) 2003-05-19 2018-05-16 Antibodies to Alpha Synuclein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47192903P 2003-05-19 2003-05-19
US60/471,929 2003-05-19

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US85057004A Continuation-In-Part 2003-05-19 2004-05-19
US10/969,335 Continuation-In-Part US7306945B2 (en) 2003-05-19 2004-10-19 Truncated fragments of alpha-synuclein in Lewy body disease
US11/194,115 Continuation-In-Part US7358331B2 (en) 2003-05-19 2005-07-29 Truncated fragments of alpha-synuclein in Lewy body disease

Publications (3)

Publication Number Publication Date
WO2005013889A2 WO2005013889A2 (fr) 2005-02-17
WO2005013889A3 WO2005013889A3 (fr) 2005-04-28
WO2005013889A9 true WO2005013889A9 (fr) 2005-06-02

Family

ID=34135040

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/015836 WO2005013889A2 (fr) 2003-05-19 2004-05-19 Fragments tronques d'alpha-synucleine dans les affections des corps de lewy

Country Status (12)

Country Link
US (1) US7306945B2 (fr)
EP (2) EP2361928B1 (fr)
JP (2) JP4820291B2 (fr)
CA (1) CA2526900C (fr)
CY (2) CY1119133T1 (fr)
DK (2) DK1633189T3 (fr)
ES (2) ES2632216T3 (fr)
HU (2) HUE035044T2 (fr)
PL (2) PL1633189T3 (fr)
PT (2) PT2361928T (fr)
SI (2) SI2361928T1 (fr)
WO (1) WO2005013889A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11844846B2 (en) 2018-05-16 2023-12-19 Emory University Styrylbenzothiazole derivatives and uses in imaging methods

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040146521A1 (en) * 1999-06-01 2004-07-29 Schenk Dale B. Prevention and treatment of synucleinopathic disease
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US20080014194A1 (en) * 2003-10-31 2008-01-17 Elan Pharmaceuticals, Inc. Prevention and Treatment of Synucleinopathic and Amyloidogenic Disease
US8506959B2 (en) * 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US9034337B2 (en) 2003-10-31 2015-05-19 Prothena Biosciences Limited Treatment and delay of outset of synucleinopathic and amyloidogenic disease
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US7358331B2 (en) * 2003-05-19 2008-04-15 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in Lewy body disease
US7550649B2 (en) 2003-10-30 2009-06-23 Taisho Pharmaceutical Co., Ltd. Transgenic non-human mammal
US7674599B2 (en) 2003-11-08 2010-03-09 Elan Pharmaceuticals, Inc. Methods of using antibodies to detect alpha-synuclein in fluid samples
MX2007001679A (es) 2004-08-09 2007-05-23 Elan Pharm Inc Prevencion y tratamiento de la enfermedad sinucleinopatica y amiloidogenica.
US20090010894A1 (en) * 2005-12-23 2009-01-08 The Parkinson's Institute Methods and systems for identifying compounds that modulate alpha-synuclein aggregation
US20070214509A1 (en) * 2005-12-23 2007-09-13 The Parkinson's Institute Methods and systems for identifying compounds that modulate alpha-synuclein aggregation
PT3067066T (pt) 2007-02-23 2019-06-17 Univ California Prevenção e tratamento da doença sinucleinopática e amiloidogénica
US8147833B2 (en) 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
CA2678963C (fr) 2007-02-23 2018-05-01 Elan Pharmaceuticals, Inc. Prevention et traitement de maladies synucleinopathiques et amyloidogeniques
AT506535B1 (de) 2008-02-22 2010-04-15 Affiris Forschungs & Entwicklungs Gmbh Vaccine enthaltend alpha-synuclein-mimotope auf basis von peptiden
CA2759118A1 (fr) * 2008-04-17 2009-10-22 Declion Pharmaceuticals, Inc. Conception et synthese de compositions de polymeres sequences dirigees et leurs anticorps pour le traitement de troubles conformationnels des proteines
PL2282758T3 (pl) 2008-04-29 2019-04-30 Bioarctic Ab Przeciwciała i szczepionki do zastosowania w terapeutycznych i diagnostycznych sposobach dla zaburzeń związanych z alfa-synukleiną
AT508638B1 (de) * 2009-08-21 2011-08-15 Affiris Ag Verwendung von peptiden und polypeptiden zur behandlung und/oder prävention von synukleinopathien
EP2635906A4 (fr) * 2010-11-05 2014-04-02 Univ Brandeis Composés inhibiteurs de ice et leurs utilisations
EP2659907A1 (fr) 2012-05-01 2013-11-06 Affiris AG Compositions
EP2659908A1 (fr) 2012-05-01 2013-11-06 Affiris AG Compositions
EP2659906A1 (fr) 2012-05-01 2013-11-06 Affiris AG Compositions
WO2013173827A2 (fr) * 2012-05-18 2013-11-21 Board Of Regents Of The University Of Nebraska Procédés et compositions pour inhiber des maladies du système nerveux central
JP2017501848A (ja) 2013-11-19 2017-01-19 プロセナ バイオサイエンシーズ リミテッド 便秘症状からのレビー小体病の免疫療法のモニター
JP2017536102A (ja) 2014-10-16 2017-12-07 ジェネンテック, インコーポレイテッド 抗アルファ−シヌクレイン抗体及び使用方法
GB201512203D0 (en) 2015-07-13 2015-08-19 Lundbeck & Co As H Agents,uses and methods
MA45125B1 (fr) 2016-06-02 2021-11-30 Medimmune Ltd Anticorps anti-alpha-synucléine et leurs utilisations
AU2017292172A1 (en) * 2016-07-06 2019-01-03 Prothena Biosciences Limited Assay for detecting total and S129 phosphorylated alpha-synuclein
MX2019005594A (es) 2016-11-15 2019-07-04 H Lundbeck As Agentes, usos y metodos para el tratamiento de la sinucleinopatia.
CN110072888B (zh) 2016-12-16 2023-07-18 H.隆德贝克有限公司 药剂、用途和方法
US10364286B2 (en) 2016-12-22 2019-07-30 H. Lundbeck A/S Monoclonal anti-alpha-synuclein antibodies for preventing tau aggregation
WO2018151821A1 (fr) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Anticorps anti-alpha-synucléine et leurs utilisations
GB201720970D0 (en) 2017-12-15 2018-01-31 Ucb Biopharma Sprl Antibodies
GB201720975D0 (en) 2017-12-15 2018-01-31 Ucb Biopharma Sprl Anti-alpha synuclein antibodies
WO2019157527A2 (fr) * 2018-02-12 2019-08-15 The Scripps Research Institute Méthodes associées à la maladie de parkinson et aux synucléinopathies
EP4192493A1 (fr) 2020-08-04 2023-06-14 AC Immune SA Composés immunogènes
KR102505164B1 (ko) * 2020-09-29 2023-02-28 서울대학교산학협력단 신경퇴행성 질환의 예방 또는 치료용 펩타이드 및 이를 포함하는 약학 조성물
WO2023041524A2 (fr) * 2021-09-16 2023-03-23 H. Lundbeck A/S Compositions et méthodes de traitement des synucléinopathies

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH652145A5 (de) 1982-01-22 1985-10-31 Sandoz Ag Verfahren zur in vitro-herstellung von hybridomen welche humane monoklonale antikoerper erzeugen.
US4634666A (en) 1984-01-06 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Human-murine hybridoma fusion partner
DE3521994A1 (de) 1985-06-20 1987-01-02 Bayer Ag N-(2-aminoacylamido-2-desoxy-hexosyl)-amide-, -carbamate und -harnstoffe, verfahren zu ihrer herstellung sowie ihre verwendung in arzneimitteln
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JPH0832638B2 (ja) 1989-05-25 1996-03-29 カイロン コーポレイション サブミクロン油滴乳剤を含んで成るアジュバント製剤
SG48759A1 (en) 1990-01-12 2002-07-23 Abgenix Inc Generation of xenogenic antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
ATE160818T1 (de) 1990-06-01 1997-12-15 Chiron Corp Zusammensetzungen und verfahren zur identifizierung von molekülen mit biologischer wirksamkeit
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
DE69230142T2 (de) 1991-05-15 2000-03-09 Cambridge Antibody Tech Verfahren zur herstellung von spezifischen bindungspaargliedern
US5858657A (en) 1992-05-15 1999-01-12 Medical Research Council Methods for producing members of specific binding pairs
JP4124480B2 (ja) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド 免疫グロブリン変異体
DE69228606T2 (de) 1991-07-03 1999-06-24 Kanebo Ltd Verfahren und vorrichtung zur herstellung eines thermoplastischen polyurethan-elastomers
WO1993006121A1 (fr) 1991-09-18 1993-04-01 Affymax Technologies N.V. Procede de synthese de diverses collections d'oligomeres
ES2136092T3 (es) 1991-09-23 1999-11-16 Medical Res Council Procedimientos para la produccion de anticuerpos humanizados.
CA2127450C (fr) 1992-01-07 2007-04-17 Samuel Wadsworth Modeles d'animaux transgeniques pour la maladie d'alzheimer
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
HUT72495A (en) 1992-10-01 1996-05-28 Cold Spring Harbor Lab Complex combinatorial chemical libraries encoded with tags
US5837242A (en) 1992-12-04 1998-11-17 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
JPH09508353A (ja) 1993-11-02 1997-08-26 アフィマックス テクノロジーズ ナムローゼ フェンノートシャップ 分子多様性の合成及びスクリーニング
US5914349A (en) 1994-01-10 1999-06-22 Teva Pharmaceutical Industries, Ltd. Compositions containing and methods of using 1-aminoindan and derivatives thereof and process for preparing optically active 1-aminoindan derivatives
AU691296B2 (en) 1994-05-06 1998-05-14 Pharmacopeia Drug Discovery, Inc. Combinatorial dihydrobenzopyran library
US5663046A (en) 1994-06-22 1997-09-02 Pharmacopeia, Inc. Synthesis of combinatorial libraries
EP1005368B1 (fr) 1997-03-10 2009-09-02 Ottawa Hospital Research Institute Utilisation d'acides nucléiques contenat un dinucléotide CpG non-methylé combiné avec de l'aluminium en tant qu'adjuvants
WO1999050300A1 (fr) * 1998-03-30 1999-10-07 The Trustees Of The University Of Pennsylvania Methode d'identification, de diagnostic et de traitement de troubles neurodegeneratifs a niveaux eleves de synucleine
US6184351B1 (en) * 1998-09-25 2001-02-06 Amgen Inc. α-synuclein super-mutants accelerate α-synuclein aggregation
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
WO2002004482A1 (fr) 2000-07-07 2002-01-17 Panacea Pharmaceuticals, Inc. Procedes pour la prevention des degats relatifs aux tissus nerveux et pour le traitement des maladies liees a l'alpha-synucleine
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11844846B2 (en) 2018-05-16 2023-12-19 Emory University Styrylbenzothiazole derivatives and uses in imaging methods

Also Published As

Publication number Publication date
PL1633189T3 (pl) 2017-12-29
EP2361928A1 (fr) 2011-08-31
CA2526900C (fr) 2015-11-24
HUE034320T2 (en) 2018-02-28
SI1633189T1 (sl) 2017-12-29
DK2361928T3 (en) 2017-07-03
EP2361928B1 (fr) 2017-04-26
US20050198694A1 (en) 2005-09-08
JP4820291B2 (ja) 2011-11-24
EP1633189B1 (fr) 2017-07-05
PL2361928T3 (pl) 2017-09-29
CY1119133T1 (el) 2018-02-14
EP1633189A4 (fr) 2006-07-19
DK1633189T3 (en) 2017-08-21
PT2361928T (pt) 2017-07-21
WO2005013889A2 (fr) 2005-02-17
US7306945B2 (en) 2007-12-11
ES2632216T3 (es) 2017-09-11
ES2640669T3 (es) 2017-11-03
WO2005013889A3 (fr) 2005-04-28
JP2011033633A (ja) 2011-02-17
SI2361928T1 (sl) 2017-07-31
JP5766923B2 (ja) 2015-08-19
PT1633189T (pt) 2017-10-04
CY1119556T1 (el) 2018-03-07
EP1633189A2 (fr) 2006-03-15
JP2007525464A (ja) 2007-09-06
HUE035044T2 (en) 2018-05-02
CA2526900A1 (fr) 2005-02-17

Similar Documents

Publication Publication Date Title
EP2361928B1 (fr) Fragments tronqués d'alpha-synucléines dans une maladie de corps de Lewy
US20180355028A1 (en) Antibodies to Alpha Synuclein
CA2503561C (fr) Prevention et traitement d'une maladie synucleopathique
DK2450056T3 (en) Prevention and treatment of synucleinopathic and amyloidogenic disease
EP1793855B2 (fr) Prevention et traitement de maladies synucleinopathique et amyloidogenique
CA2912912C (fr) Fragments tronques d'alpha-synucleine dans les affections a corps de lewy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
COP Corrected version of pamphlet

Free format text: PAGES 8/11 AND 11/11, DRAWINGS, REPLACED BY NEW PAGES 8/11 AND 11/11; AFTER RECTIFICATION OF OBVIOUS ERRORS AUTHORIZED BY THE INTERNATIONAL SEARCH AUTHORITY

WWE Wipo information: entry into national phase

Ref document number: 11194115

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2006514909

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2526900

Country of ref document: CA

REEP Request for entry into the european phase

Ref document number: 2004776059

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2004776059

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004776059

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11194115

Country of ref document: US